WO2014068001A1 - Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain - Google Patents

Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain Download PDF

Info

Publication number
WO2014068001A1
WO2014068001A1 PCT/EP2013/072717 EP2013072717W WO2014068001A1 WO 2014068001 A1 WO2014068001 A1 WO 2014068001A1 EP 2013072717 W EP2013072717 W EP 2013072717W WO 2014068001 A1 WO2014068001 A1 WO 2014068001A1
Authority
WO
WIPO (PCT)
Prior art keywords
proteins
cmv
capsid
towne
protein
Prior art date
Application number
PCT/EP2013/072717
Other languages
English (en)
Inventor
Sabine Wellnitz
Corinne John
Christian Schaub
Original Assignee
Redbiotec Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to ES13789738.5T priority Critical patent/ES2608637T3/es
Priority to RU2015121828A priority patent/RU2015121828A/ru
Application filed by Redbiotec Ag filed Critical Redbiotec Ag
Priority to SG11201501623PA priority patent/SG11201501623PA/en
Priority to BR112015008930A priority patent/BR112015008930A2/pt
Priority to CN201380057084.6A priority patent/CN104853772A/zh
Priority to AU2013340820A priority patent/AU2013340820A1/en
Priority to KR1020157013906A priority patent/KR20150076244A/ko
Priority to CA2885145A priority patent/CA2885145A1/fr
Priority to EP13789738.5A priority patent/EP2914284B1/fr
Priority to JP2015540112A priority patent/JP6294890B2/ja
Priority to MX2015005505A priority patent/MX2015005505A/es
Priority to US14/436,943 priority patent/US20150359879A1/en
Publication of WO2014068001A1 publication Critical patent/WO2014068001A1/fr
Priority to IL237793A priority patent/IL237793A0/en
Priority to PH12015500931A priority patent/PH12015500931A1/en
Priority to HK16101176.6A priority patent/HK1213185A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16123Virus like particles [VLP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the invention relates to virus-like particles comprising capsid proteins, cytomegalovirus surface proteins, and optionally tegument proteins, and vaccines against human cytomegalovirus infection.
  • HCMV Human cytomegalovirus
  • antiviral reagents such as ganciclovir (valganciclovir), foscarnet, acyclovir, cidofovir or leflunomide.
  • ganciclovir valganciclovir
  • foscarnet acyclovir
  • cidofovir leflunomide
  • antiviral agents have been approved for transplant recipients and immunocompromised patients (e.g. HIV patients), but not in pregnant women. For these a hyper- immunoglobin therapy is preferred.
  • the existing antiviral therapies lead to viral resistance in a short timeframe or were not effective during therapy (McGregor A. et al., Expert Opin. Drug Metab. Toxicol. 201 1 ; 7(10): 1245-1265).
  • Virus-like particles can accommodate properties similar to attenuated viruses due to their shape and are therefore a promising option for vaccine development, especially since they are generally accepted as excellent vaccine vehicles. Virus-like particles have not been developed for CMV up to now.
  • the following goals should be met: a long-lasting induction of neutralizing antibodies, induction of humoral and cellular, especially cytotoxic T-cell responses and minimizing side effects.
  • the reasons for failure or limited efficacy of vaccine candidates developed so far are multifaceted.
  • the mechanisms of protective immunity in a host are poorly elucidated.
  • One of the main reasons for this is the lack of an appropriate animal model to be able to verify the effectors accounting for protection.
  • CMV is a beta-herpesvirus and belongs to the very complex viral order Herpesvirales.
  • the CMV virion is -230 nm in diameter and is composed of a nucleocapsid, surrounded by a less structured tegument layer, and bounded by a trilaminate membrane envelope.
  • the linear, double-stranded DNA molecule (236 kbp) is the largest among the human herpesviruses and over 50% larger than that of Herpes simplex virus 1 (HSV-1 ).
  • HSV-1 Herpes simplex virus 1
  • DB dense bodies
  • DBs Dense bodies
  • DBs are large (-250-600 nm) and heterogeneous. They are solid spheroidal aggregates of different proteins containing a tegument protein (i.e.
  • gCI- gCIII three major complexes (designated as gCI- gCIII) have been identified so far. These complexes play different roles in the virus structure and thus also for the design of vaccines, as targets for the development of therapeutic antibodies, or as research tools.
  • the glycoprotein B (gpUL55, gCI) as a single soluble protein did not lead to a long lasting immune response against HCMV. Therefore, further protein compositions including gpUL55 should be taken into account for vaccine design.
  • the role of gpUL100 (gM), gpUL73 (gN) and the formed complex gCII for generation of neutralizing antibodies and as an essential part of a vaccine is under investigation.
  • gCIII Constituents of gCIII are gpUL75 (gH), gpUL1 15 (gl_) and gpUL74 (gO) forming a heterotrimeric, disulfide-linked complex (Huber M.T. and Compton T., J. Virol. 1999; 73(5):3886-3892). These proteins play the most important role for viral entry and complex formation with further proteins such as gpUL128, gpUL130 and gpUL131A.
  • HCMV The entry of HCMV into epithelial and endothelial cells involves endocytosis and complex formation of gpUL75, gpUL1 15 and gpUL74, or of the combination of gpUL75, gpUL1 15, gpUL128, gpUL130 and gpUL131 (Wang D. and Shenk T., PNAS 2005; 102:18153- 18158).
  • these proteins are responsible for the transfer of HCMV from endothelial cells to leukocytes, representing major determinants of HCMV dissemination in vivo.
  • these proteins seem to play a role in the priming of dendritic cells (DC) and are therefore critical elements for T-cell mediated immune response.
  • DC dendritic cells
  • gpUL75/gpUL1 15 gH/gL
  • gpUL75/gpUL1 15/gpUL74 gH/gL/gO
  • the second important pathway is an endosomal infection process of epithelial and endothelial cells which requires the pentameric complex gpUL75/ gpUL1 15/ gpUL128/ gpUL130/ gpUL131 (gH/gL/UL128/UL130/UL131A).
  • This infection route can be blocked by neutralizing antibodies targeting the pentameric complex (Manley K. et al., Cell Host & Microbe 201 1 ; 10:197-209). From the current state of knowledge different combinations of the above proteins are required to generate an efficient vaccine that stimulates a broad cellular and humoral immune response. However, the exact combination is not known.
  • the importance of the proteins of the pentameric complex was addressed recently by Lilleri D. et al., PLoS One, March 2013 ; 8(3): e59863, 1 -13.
  • VLPs Virus-like particles
  • Virus-like particles are stable, highly organised spheres that self-assemble from virus-derived structural antigens and thus have structural characteristics and antigenicity similar to the parental virus.
  • the recombinant VLPs are safe and non-infectious since they do not contain replicating viral DNA. Therefore, they can be generated under biosafety level 1 .
  • VLPs provide, on the basis of their particulate nature, an inherent advantage over soluble antigens in respect to immunogenicity and stability. Compared to subunit or single recombinant proteins, VLPs are more immunogenic and are able to stimulate B-cell mediated immune responses as well as CD4 proliferative and CTL responses (Ludwig C. and Wagner R., Curr. Opin. Biotechnol. 2007; 18:537-545). The size of VLPs appears to be favourable for uptake by dentritic cells (DC) through endocytosis or macropinocytosis and activating innate and adaptive immune response. VLPs, especially retroviral VLPs (gag-VLPs), contain immunogenic epitopes and are supposed to stimulate cellular immune responses via MHC class-l and MHC class-ll pathways (Demi L. et al., Mol.
  • VLPs do not appear to require the use of adjuvants to achieve potent immune stimulation.
  • VLPs have been extensively, and quite successfully, used as vaccine (WO 2005/123125) and viral serology reagents.
  • VLPs For enveloped viruses, VLPs require at least one capsid or matrix protein for the assembly of a viral particle. The proteins assemble in different cellular compartments such as endoplasmic reticulum (ER), lipid rafts or plasma membrane where the budding takes place, and thus contain the cellular lipids building the viral lipoprotein envelope.
  • the VLPs may also include host cell proteins, e.g. lipid raft associated gangliosides.
  • VLPs can be exploited for presentation of foreign epitopes and/or targeting molecules based on single or multicomponent capsids.
  • the core gene of hepatitis B virus fused with antigens provided an early example of the VLP approach (WO 1999/057289).
  • no recombinant CMV VLP has been generated.
  • the largest VLPs generated and published are Influenza, Bluetongue and Rotavirus VLPs that contain up to 5 different proteins. It is not exactly known how many and which proteins are necessary to form a recombinant CMV particle based on CMV capsid proteins. In contrast, many non-herpesvirus capsid proteins are known to form VLPs.
  • the most prominent example is the retroviral precursor protein gag, in particular MoMuLV (Moloney Murine Leukemia Virus), HIV (Human Immunodeficiency virus), HTLV (Human T-Cell Leukemia Virus) and SIV (Simian
  • the pr55 HIV precursor protein forms a VLP with a size of 100-120 nm and was shown to serve as carrier for further proteins
  • the MoMuLVprecursor protein gag (pr65) leads to VLPs with a size of 1 15-150 nm in a high yield based on immunoblotting and ELISA assays.
  • BEVS baculovirus expression vector system
  • the primary production host are insect cells, however, the baculovirus can also be applied for recombinant co-expression in mammalian cells.
  • the baculovirus system is a widely used and highly efficient system for protein expression in research and commercial laboratories around the world (Roldao A. et al., Expert Rev. Vaccines, 2010; 9(10):1 149- 1 176).
  • GlaxoSmithKline's human papillomavirus (HPV) VLP vaccine is produced in BEVS and approved for use in the USA and the European Union (WO 2005/123125).
  • WO 2010/128338 discloses CMV vaccines comprising Herpes Simplex Virus (HSV) or a HSV virus-like particle (HSV VLP) further comprising a Human Cytomegalovirus (HCMV) polyepitope. Summary of the Invention
  • the invention relates to a recombinant virus-like particle comprising one or more capsid or capsid precursor proteins, 3, 4, 5 or more different surface proteins from cytomegalovirus (CMV), and optionally one or more tegument proteins.
  • CMV cytomegalovirus
  • the capsid proteins are derived from a herpes virus such as cytomegalovirus, e.g. human cytomegalovirus (HCMV), or from a retrovirus, in particular HIV or MoMuLV.
  • cytomegalovirus e.g. human cytomegalovirus (HCMV)
  • HCMV human cytomegalovirus
  • retrovirus in particular HIV or MoMuLV.
  • Preferred capsid protein is the HIV and/or MoMuLV precursor gag protein.
  • the surface proteins are preferably selected from HCMV, in particular from the group consisting of gpUL75 (gH), gpUL1 15 (gl_), gpUL55 (gB), gpUL74 (gO), gp100 (gM), gp73 (gN), gpUL128, gpUL130, and gpUL131A.
  • the tegument proteins are preferably selected from HCMV, in particular from the group consisting of pUL32, pUL45, pUL47, pUL48, pUL69, pUL71 , pUL72, pUL76, pUL77, pUL83 (pp65), pUL88, pUL93, pUL94, pUL95, pUL97, pUL99, and pUL103.
  • the core proteins are in particular selected from the group consisting of pUL23, pUL24, pUL33, pUL36, pUL38, pUL43, pUL78, pUL82, pUL96, IRS1 , US22, and TRS1 having either tegument or envelope protein character.
  • the regulatory proteins are preferably selected from the group of IE-1 , UL50, UL80.5, UL46 and UL47.
  • the recombinant virus-like particle according to the invention may further comprise B- and/or T-cell epitopes, proteins selected from the group consisting of additional foreign antigenic sequences, cytokines, CpG motifs, g-CMSF, CD19, and CD40 ligand, and/or fluorescent proteins, proteins useful for purification purposes of the particles or for attaching a label, and/or proteinaceous structures required for transport processes.
  • the invention relates to a DNA encoding the proteins comprised in the viruslike particles according to the invention, to a vector comprising such DNA, in particular a baculovirus vector, and a host cell comprising such a vector, and to methods of manufacturing virus-like particles according to the invention using a baculovirus vector.
  • the invention relates to a vaccine comprising a recombinant virus-like particle according to the invention, in particular to such a vaccine further comprising the pentameric complex consisting of gpUL75, gpUL1 15, gpUL128, gpUL130 and gpUL131A, and/or soluble CMV protein selected from the group consisting of gpUL75, gpUL1 15, gpUL.55, gpUL.74, gpUL.100, and gpUL73; or gpUL128, gpUL.130, and gpUL131A; or pUL83, IE-1 , UL99, UL91 , and pp150.
  • Such a vaccine may further comprise an adjuvant selected from the group consisting of aluminium hydroxide, alum, AS01 , AS02, AS03, AS04, MF59, MPL, QS21 , ISCOMs, IC31 , unmethylated CpG, ADVAX, RNA containing formulations and Freund's reagent, but also DNA of the invention as defined above.
  • an adjuvant selected from the group consisting of aluminium hydroxide, alum, AS01 , AS02, AS03, AS04, MF59, MPL, QS21 , ISCOMs, IC31 , unmethylated CpG, ADVAX, RNA containing formulations and Freund's reagent, but also DNA of the invention as defined above.
  • the vaccine comprises CMV proteins from different CMV strains selected from the group of Towne, Toledo, AD169, Merlin, TB20, and VR1814 strains.
  • the vaccine comprising a recombinant virus-like particle according to the invention may be further enhanced in its induction of host immune response by a second immunization with the pentameric complex consisting of gpUL75, gpUL1 15, gpUL128, gpUL130, and gpUL131A in a prime-boost administration, and/or with a soluble CMV protein selected from the group consisting of gpUL75, gpUL1 15, gpUL55, gpUL74, gpULI OO, gpUL73, pUL83, and IE-1 in a prime-boost administration.
  • the vaccine comprising a recombinant virus-like particle according to the invention may be further enhanced in its induction of host immune response (humoral and cellular response) by addition of the soluble complexes consisting of at least two different surface proteins out of the group consisting of gpUL73, gpUL74, gpUL75, gpULI OO, gpUL1 15, gpUL128, gpUL130, and gpUL131A.
  • Figure 1 Schematic representations of recombinant vectors for expression of different variants of CMV virus-like particles, either based on a herpesvirus or non-herpesvirus capsid, or for expression of CMV-pentameric complex and soluble CMV proteins.
  • the different variants are inserted into the vector backbone pRBT136 aimed at recombinant protein expression using the baculovirus expression system (BEVS) and containing two promoters P1 and P2 ( ⁇ l_p10, _polh) and two terminator sequences T1 and T2 (T), which are SV40 and HSVtk.
  • BEVS baculovirus expression system
  • T1 and T2 T1 and T2
  • T SV40 and HSVtk
  • the vectors contain an origin of replication (O), e.g. 2micron, and a marker gene (m), e.g. URA3.
  • O origin of replication
  • m marker gene
  • the vectors contain the transposon sites left (TL) and right (TR) for transposition of the transgenes from the transfer vector into bacmids, a loxP site (L) for site specific homologous recombination (plasmid fusion), origins of replication (O), ampicillin (A), chloramphenicol (C) and gentamycin (G) resistance genes, and defined restriction sites.
  • the vector backbone pRBT 393 contains in addition a promoter selected from pCMV, ie1 and Ief2, and a terminator selected from SV40pA, BHG pA and HSVtk.
  • Figure 2 Glycerol-tartrate gradient based purification of a CMV-VLP variant SEQ ID NO:7 combined with SEQ ID NO: 14.
  • Number 1 -13 of the x-axis represents the number of the glycerol-tartrate gradient (fractionated from top to bottom), number 14 represents a positive and number 15 a negative control. Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs. Similar results were obtained for the expression and purification of the variants SEQ ID NO:7 combined with SEQ ID NO:9, SEQ ID NO:10 and SEQ ID NO:13.
  • Figure 3 Glycerol-tartrate gradient based purification of a CMV-VLP variant SEQ ID N0:5 combined with SEQ ID NO: 14.
  • Number 1 -13 of the x-axis represents the number of the glycerol-tartrate gradient (fractionated from top to bottom), number 14 represents a positive and number 15 a negative control. Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs.
  • Figure 4 Glycerol-tartrate gradient based purification of a CMV-VLP variant SEQ ID NO:3 combined with SEQ ID NO:18.
  • Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs. The same intact composition was observed by combining SEQ ID NO:3 with SEQ ID NO:9, 10, 16 or 19 where the OD value detection differs in a range between 0.1 - 0.4.
  • Figure 5 Glycerol-tartrate gradient based purification of a CMV-VLP variant SEQ ID N0:2 combined with SEQ ID N0:18.
  • Number 1 of the x-axis represents the sample of the pre-purification by sucrose cushion
  • lanes 2-14 of the x-axis represents the number of the glycerol-tartrate gradient fractions 1 to 13 (fractionated from top to bottom)
  • number 15 represents a negative control for VLPs
  • number 16 a negative control for baculoviruses.
  • Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs.
  • Figure 6 Sucrose gradient based purification of non-herpesvirus capsid based CMV-VLP variant SEQ ID NO:20 combined with SEQ ID NO:14.
  • Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs. The same co-localization could be observed by combining the capsid protein gag (SEQ ID NO:20) with the tegument and surface proteins of SEQ ID NO: 9, 10, 13, 15,16 or 19. The inclusion of SEQ ID NO: 19 showed in general higher OD values indicating better stability of the CMV VLP.
  • Figure 7 Sucrose gradient based purification of non-herpesvirus capsid based CMV-VLP variant SEQ ID NO: 20 combined with SEQ ID NO: 18.
  • Number 1 -13 of the x-axis represents the number of the sucrose gradient fractions 1 to 13 (fractionated from top to bottom), number 14 represents the load of the gradient and number 15 a positive and number 16 a negative control.
  • Binding strength of antibody to protein in the VLPs is shown as optical density (OD) at the y-axis. Detection of the selected proteins in the same fractions showed their co-localization and therefore intact CMV-VLPs.
  • a further CMV variant based on the expression of SEQ ID NO:22 and SEQ ID NO:18 led to similar results with the benefit to support the induction of cellular immune response by the protein UL83.
  • Figure 8 Analysis of the His/Strep-tagged CMV-VLP variant SEQ ID NO:7 combined with SEQ ID NO:14 purified by affinity chromatography.
  • the CMV-VLP variant comprising capsid, functional, tegument and surface proteins UL86- UL85-UL50-UL48A-UL46-UL74-UL83-UL80.5-UL75-UL1 15(His/Strep)-UL128(c)-UL130- UL131A (SEQ ID NO:7 and SEQ ID NO:14) was purified with an IMAC procedure.
  • UL83 antibody (Virusys).
  • Lane 1 VLP before purification (load)
  • lane 2 flowthrough (FT)
  • lane 3 wash
  • lanes 4 to 9 elution with increasing amount of imidazole (55, 100, 150, 230, 480, 500 mM)
  • lane 10 insect cells (Sf9) as negative control
  • lane 1 1 positive control (infected Sf9 cells). Presence of the tegument protein indicated by an arrow (UL83).
  • Figure 9 Analysis of the His/Strep-tagged non-herpesvirus capsid based CMV-VLP variant SEQ ID NO:20 combined with SEQ ID NO:14 purified by affinity chromatography.
  • the CMV-VLP variant comprising the capsid (retroviral precursor) protein gag and surface proteins UL75-UL1 15(His/Strep)-UL128(c)-UL130-UL131A (SEQ ID NO:20 and SEQ ID NO:14) was purified with an IMAC based procedure. Analysis of different steps of this
  • FT flowthrough
  • lane 3 wash
  • lane 4 to 9 elution with increasing amount of imidazole (55, 100, 150, 230, 480, 500 mM)
  • lane 10 insect cells (Sf9) as negative control
  • lane 1 1 positive control (infected Sf9 cells).
  • Figure 10 Analysis of purification process consisting of two affinity chromatography steps, followed by size exclusion chromatography of the His-tagged soluble CMV-pentameric complex.
  • the pentameric complex comprising the surface proteins UL75 (His-tagged, gH)-UL1 15 (gL)-UL128-UL130-UL131A (SEQ ID NO: 59) was purified by an affinity based
  • Lane 1 represents the elution pool IV-2 (precipitated); lane 2: elution pool IV-2 (non precipitated); lane 3: elution pool IV- 3; lane 4: elution pool V; lane 5: elution pool VI; lane 6: positive control.
  • the His-tagged gH protein is marked on the right side.
  • Figure 11 Electron micrograph of HCMV-VLP variant SEQ ID NO:7 and SEQ ID NO:10 (A) and SEQ ID NO:48 and SEQ ID NO: 19 (B).
  • Figure 12 Electron micrograph of non-herpes virus based CMV-VLP variant SEQ ID NO:20 and SEQ ID NO:10 (A) and SEQ ID NO:88 and SEQ ID NO:19 (B).
  • Figure 13 Virus neutralization assay with mouse sera based on in vivo studies with a vaccine combination of SEQ ID NO:59 and SEQ ID NO:22 with SEQ ID NO: 18, respectively.
  • Balb/C mice were immunized in a prime-boost-boost regimen with hCMV antigens.
  • the selected antigen for this neutralization assay is a protein mix of the soluble complex (SEQ ID NO:59) and the VLP composed of a retroviral capsid in combination with the tegument protein UL83 (SEQ ID NO:22) and the CMV proteins UL55-UL75-UL1 15-UL128-UL130 and UL131A.
  • Pooled sera obtained from eight mice in a two-fold serial dilution (1 :20 to 1 :2560) were mixed with a recombinant, BAC-reconstituted VR1814 HCMV strain expressing EGFP under the CMV promoter.
  • MRC-5 fibroblasts (2x10 4 cells/well) were used to assess the entry of this virus in a cell-based fluorescence assay, 96 h post treatment.
  • the relative fluorescence intensities (RFI, %) are depicted for selected sera dilutions from mice immunized with a mixture of non-herpes CMV-VLP and the pentameric CMV complex. An empty baculovirus and PBS were used as control.
  • the immunisation with a mix of soluble pentameric complex and a non-herpesvirus based CMV VLP induced the generation of neutralising antibodies within a titer range of 1 :160 to 1 :320 comparable with titers of human blood donors using the pre-evaluated TCID 50 for the used EGFP- virus.
  • Figure 14 Neutralization assay to verify humoral immune response based on SEQ ID NO:59
  • PR pentamer pre-immune
  • PO pentamer post-immune
  • G1 negative blood donors
  • G2 positive blood donors.
  • Figure 15 Quality control of one component (soluble complex, SEQ ID NO: 59) of the vaccine candidate containing the soluble complex and reVLP (recombinant VLP, SEQ ID NO:22 with SEQ ID NO: 18).
  • Figure 16 Quality control of one component (soluble complex, SEQ ID NO: 59) of the vaccine candidate containing the soluble complex and the reVLP (recombinant VLP, SEQ ID NO:22/18) using conformation-dependent antibodies.
  • Figure 17 Validation of a non-herpes-based CMV-VLP (SEQ ID NO: 22 combined with SEQ ID NO: 18) with sandwich ELISA assay.
  • Figure 18 Characterization of a purification process of a non-herpes-based CMV-VLP (SEQ ID NO: 22 combined with SEQ ID NO: 18) with a sandwich ELISA assay.
  • Elution fractions from two different chromatography matrices were tested for the co-presence of the designated proteins.
  • Samples were captured with a conformation-dependent (UL130/UL131A) antibody and detected with an anti-gH, an anti-pp65, an anti-gB and an anti-gag (capsid) antibody.
  • the signals confirm co-existence of the proteins on a VLP, and reveal the better performance of the membrane adsorber (c2) used.
  • 1 elution fraction from column 1 (c1 )
  • 2 elution fraction from column 2 (c2)
  • 3 positive control
  • 4 negative control.
  • Cap capsid (precursor gag protein).
  • Figure 19 Characterization of a herpes-based CMV-VLP with sandwich ELISA assay.
  • VLPs were produced with two ( Figure 19A, UL86 and UL80.5, SEQ ID NO:2) or three ( Figure 19B, UL86-UL85-UL80.5, SEQ ID NO:3) different capsid proteins; the same tegument protein UL83 (pp65) and different surface proteins (gH-gL-gB-UL128-UL130- UL131A, SEQ ID NO:18), and elution fractions from the subsequent purification step were subjected to sandwich ELISA.
  • FIG. 20 Cellular immune response induced by a combination of a non-herpes CMV- VLP (SEQ ID NO:22/SEQ ID NO: 18) and the pentameric CMV complex (SEQ ID NO:59).
  • Spleen cells were re-stimulated 24 h post harvesting with various peptides (1 -7), and the release of the Th1 -type cytokines, IFN-g (A) and IL-2 (B), the Th2-type cytokines, IL-4 (C), IL-5 (D) and IL-10 (E), and the inflammatory cytokines, GM-CSF (F) and TNFa (G) was measured with a multiplex assay according to manufacturer's protocol (Invitrogen).
  • Peptides 2-5 were a mixture of nonamers with predicted epitopes (SYFPEITHI program), whereas peptides 6 and 7 were commercially purchased (JPT, mix of pre-defined peptides). An empty baculovirus and PBS (mock) were used as controls for the re- stimulation.
  • Figure 21 Analysis of CMV VLPs and single components (soluble complex, SEQ ID NO:59) for a vaccine using human CMV positive sera.
  • Non-herpes virus based CMV VLP (SEQ ID NO:22 combined with SEQ ID NO:19), herpes virus based CMV VLPs (SEQ ID NO:48 combined with SEQ ID NO:19) and the soluble complex (SEQ ID NO:59) were tested in a sandwich ELISA assay for their ability to react with human antibodies from CMV-positive donors to simulate an in vivo experiment.
  • Plates were coated with antibodies corresponding to different CMV proteins (gH, gB, UL83, UL85, UL86 and conformation-based UL130/UL131A), to Gag and to His. The plates were further incubated with either the soluble pentameric complex or VLPs and subsequently treated with CMV-positive human sera (HIV negative donors). An HRP- labeled secondary human anti-lgG was used for detection.
  • CMV proteins gH, gB, UL83, UL85, UL86 and conformation-based UL130/UL131A
  • a CMV-based VLP UL86-UL85-UL83-UL80.5-UL74-gL-gH-UL128-UL130-UL131A-gB- UL83 (SEQ-ID NO: 48 combined with SEQ-ID NO:19) was analyzed with human sera in a sandwich ELISA assay. All proteins in these VLPs were recognized by human antibodies in donors' sera with very clear signals for UL83, gH, UL85, UL86. 1 : Gag-based CMV VLP; 2: negative control (mock). The remaining baculovirus in the CMV-VLP and soluble complex manufacturing did not bind to the antibodies in present in human sera.
  • the invention relates to novel virus-like particles for use as anti-HCMV vaccine, for development of therapeutic antibodies, in anti-HCMV treatment, as diagnostic tools and as R&D tools.
  • the present invention describes several possibilities to generate
  • VLPs multicomponent VLP variants and combinations of different protein compositions such as VLPs, protein complexes, soluble proteins and/or DNA based compositions such as vectors and peptides.
  • the recombinant VLPs of the present invention are based on a capsid formed by either a CMV capsid protein combination of 1 to 6 proteins (major capsid, minor capsid, smallest capsid protein and necessary proteins for assembly) or on a retroviral precursor protein, in particular the one of the lentiviral human immunodeficiency virus (HIV) named gag, and/or pr55. Further proteins are added to these capsid proteins selected from CMV tegument proteins and surface proteins.
  • the VLP formed is surrounded by an envelope provided by the host cell during the secretion process. In order to connect and/or embed the tegument and surface proteins in their natural conformation, their individual membrane anchors are present.
  • a CMV virus contains at least 5 capsid proteins (gene products of UL46, UL48A, UL85, UL86, UL104), 19 regulatory proteins, 17 tegument proteins (gene products of UL25, UL45, UL47, UL48, UL69, UL71 , UL72, UL76, UL77, UL83, UL88, UL93, UL94, UL95, UL97, UL99, UL103), 5 surface or envelope proteins (gene products of UL55 [gB], UL73 [gN], U74 [gO], UL75 [gH], UL100 [gM], UL1 15 [gl_]), the non-categorized gene products from the open reading frame UL128, UL130, UL131A; proteins from 15 beta- herpesvirus specific genes (UL23, UL24, UL32, UL33, UL35, UL36, UL38, UL43, UL74 [gO], UL
  • the invention relates to a recombinant virus-like particle comprising one or more capsid or capsid precursor proteins, 3 or more different surface proteins from cytomegalovirus (CMV), and optionally one or more tegument proteins.
  • CMV cytomegalovirus
  • the surface proteins are selected from the group consisting of pUL83 (pp65), gpUL75 (gH), gpUL55 (gB), gpUL100 (gM), gpUL73 (gN), gpUL74 (gO), gpUL1 15 (gL), gpUL75 (gH), gpUL128, gpUL130, and gpUL131A, preferably from human CMV. Nevertheless proteins of non-human CMV strains may be used. Such non-human CMV proteins then may take a structural rather than an immunological function in the VLP.
  • the recombinant VLPs according to the invention may comprise proteins from other herpesvirus families such as HSV (herpes simplex virus), EBV (Epstein-Barr virus) and KSHV (Kaposi's sarcoma-associated herpesvirus).
  • HSV herpes simplex virus
  • EBV Epstein-Barr virus
  • KSHV Kaposi's sarcoma-associated herpesvirus
  • the VLPs based on CMV capsid proteins are composed of one protein or of combinations of proteins selected from the six proteins pUL86, pUL85, pUL48A, pUL46, pUL50, and pUL80.5, for example two, three, four, five, or six of these proteins.
  • the VLP comprises protein pUL104.
  • the non-herpesvirus capsid is chosen from the retroviridae out of the retroviral families, alpha-, beta-, gamma- , delta and epsilon retrovirus, the lentivirus family or the
  • avian (SIV), feline (FIV), bovine (BIV) and the human (HIV) precursor protein gag (group-specific-antigens) is used as capsid for the generation of non-herpesvirus based VLPs as well as the precursor protein gag from the gamma retrovirus, like feline leukemia virus (FELV), moloney mouse leukemia virus (Mo-MuLV), avian leukemia virus (SLV) or the murine sarcoma virus (MSV).
  • FELV feline leukemia virus
  • Mo-MuLV moloney mouse leukemia virus
  • SLV avian leukemia virus
  • MSV murine sarcoma virus
  • the VLPs based on a non-herpesvirus capsid are preferably composed of the HIV precursor protein gag (pr55), which is a precursor protein composed of a matrix (MA, p17), a capsid (CA, p24), a nucleocapsid (NC, p7) and a link (LI, p6) protein part.
  • the MoMULV gag variant which is composed of the same subunits, is suitable as retroviral based capsid carrying more than three different CMV tegument and/or surface proteins for the formation of a CMV VLP, as well as having advantages in the manufacturing and regulatory approval of a vaccine.
  • the assembly of CMV VLP variants based on the MoMuLV gag precursor protein is similar to the assembly of HIV gag based VLPs. There is no big advantage in respect to yield, better shape and conformation of the MoMULV gag based CMV-VLPs.
  • a HIV based CMV VLP human vaccine no false positive results in a diagnosis test for HIV are expected for a CMV-VLP vaccine based on a MoMuLV gag capsid.
  • a preferred VLP comprises, on top of the respective capsid proteins, one or two tegument proteins selected from the group consisting of UL83, UL25, UL32 (pp150), and UL99, and five to eight surface proteins.
  • the surface proteins are preferably selected from the group consisting of UL75 (gH), UL1 15, (gL), UL74 (gO), UL55 (gB), UL128, UL130, and UL131AA.
  • the VLP may also contain UL100 (gM) and UL73 (gN).
  • the surface proteins of the HCMV in the VLPs of the invention are selected together with their respective membrane anchor such that they are displayed and embedded in their natural conformation into the recombinant capsid structure.
  • the CMV capsid contains only the major capsid protein (UL86). In all other embodiments the capsid consists of more than one protein.
  • the VLP contains pUL86-pUL85-pUL80.5 in combination with tegument proteins pUL83 and ppUL32 (pp150) and the surface proteins gpUL75 (gH), gpUL1 15 (gL), gpUL55 (gB), gpUL128, gpUL130, and gpUL131A.
  • tegument and surface proteins are expressed on a capsid composed of pUL86-pUL85-pUL48A improving the core and therefore the VLP shape.
  • the VLP additionally comprises surface protein gpUL74 (gO). This enhances particle formation and enlarges the amount of proteins inducing protective immune response.
  • gO surface protein
  • the capsid and tegument proteins are from another herpesvirus, such as herpes simplex virus, Epstein-Barr virus, HHV-6 (roseolovirus) or HHV-7 (pityriasis rosea).
  • herpes simplex virus Epstein-Barr virus
  • HHV-6 roseolovirus
  • HHV-7 HHV-7 (pityriasis rosea).
  • the VLPs comprise at least one tegument protein and/or one surface protein of non-human CMV origin.
  • the VLPs comprise at least two different proteins fused to a peptide comprising a coiled-coiled motif, having a zipper function and connecting said at least two proteins carrying the corresponding motif.
  • Preferred proteins fused to the peptide comprising a coiled-coiled motif are one of the capsid proteins, preferably UL86, and one of the surface proteins, preferably to gpUL75, to support the assembly into a functional VLP and to increase the amount of immunogenic proteins on the surface.
  • the proteins for the generation of a multicomponent recombinant VLP are chosen from a single CMV strain, preferably selected from Towne, Toledo, AD169, Merlin, VR1814 strain, and/or clinical isolates.
  • the proteins for the generation of the VLPs are selected from different CMV strains, preferably from Towne, Toledo, AD169, Merlin, VR1814 strains and/or different clinical isolates.
  • VLPs comprising protein sequences from different CMV strains confer protection against several strains and prevent re-infection and/or re-activation with similar strains.
  • Different CMV strains considered are Towne, Toledo, AD169, Merlin, TB20, and VR1814 strains, but also other clinical isolates.
  • Towne and VR1814 combinations of protein sequences in particular in a VLP composed of gpUL55, gpUL74, gpUL75, gpUL1 15, UL128, UL130, and UL131A, and either CMV capsid protein(s) or retroviral gag capsid protein.
  • the protein compositions of the invention contain immunogenic proteins and therefore a number of different epitopes, preferably of the kind that stimulate humoral and cellular immune responses.
  • the activation of humoral immune responses by epitopes leads to the production of antibodies that will bind to proteins containing such epitopes.
  • the activation of cellular immune responses by epitopes leads to the production of cytotoxic T cells (also known as T c , CTL, T-Killer cell, cytolytic T cell, CD8+ T-cell, or killer T cell) which kill cells presenting such epitopes on their surface.
  • cytotoxic T cells also known as T c , CTL, T-Killer cell, cytolytic T cell, CD8+ T-cell, or killer T cell
  • Epitopes as understood herein may be repetitive, and may be part of a larger protein, in particular part of an antigen.
  • Different proteins comprising epitopes of the invention are, for example, gpUL75 (gH), gpUL1 15 (gL), UL128, UL130, UL131A, gpUL55 (gB), gpUL74 (gO), gpULI OO (gM), gpUL73 (gN), pUL86, pUL85, pUL80.5, pUL82, pUL83, pUL46, pUL48A, pUL50, pUL32, and the immediate early protein IE-1.
  • the VLPs comprise epitopes from a single or from different HCMV strains and/or non-human CMV strains, combined with B- and/or T-cell epitopes in order to induce a broader immune response.
  • VLPs comprising an inclusion of CD ligands e.g. CD40L, CD19L
  • Such VLPs induce the immune response via a distinct pathway resulting in a Th1 or Th2 answer.
  • VLPs comprising CpG motives, cytokines, and/or betaglucans
  • Such VLPs directly trigger B-cell activation, and enhance and broaden the immune response caused by the specific HCMV epitopes.
  • virus-like particle consists of proteins forming a complete virus-like surface, optionally further comprising a viral capsid protein.
  • the virus- like particle of the invention may further comprise fluorescent proteins, proteins useful for purification purposes of the particles or for attaching a label, and proteinaceous structures required for transport processes.
  • the invention relates to a recombinant virus-like particle comprising multiple combinations of capsid, optionally tegument and/or surface proteins from non-human and/or human cytomegalovirus in different amounts.
  • These particles contain at least a capsid protein, preferably the major capsid protein UL86, a tegument protein selected from the group of ppUL 83, ppUL32, pUL25, and the IE-1 protein, and a surface protein selected from the group consisting of gpUL75 (gH), gpUL1 15 (gL), gpUL128, gpUL130, gpUL131A, gpUL55 (gB), gpUL74 (gO), gpULI OO (gM), and gpUL73 (gN).
  • the surface proteins are preferably of human viral origin.
  • the virus-like particles of this invention consist either of one or more, such as one to sixteen proteins comprising various proteins selected either (a) from one HCMV strain or (b) from different HCMV strains and/or (c) from non-human CMV like mouse or guinea pig CMV.
  • Preferred are recombinant virus-like particles comprising one or more, preferably two or more different proteins from a single HCMV strain or different proteins from different CMV strains.
  • recombinant virus-like particles comprising one or more capsid proteins, the tegument proteins pUL83 and ppUL32, and the surface proteins gpUL55 (gB), gpUL74 (gO) and/or the proteins forming a pentameric structure selected from the group consisting of gpUL75 (gH), gpUL1 15 (gl_), gpUL128, gpUL130, and gpUL131A.
  • the recombinant virus-like particles of this invention may also be based on a non-herpesvirus capsid, preferably on the lentiviral precursor protein gag, and further be composed of CMV surface proteins and optionally CMV tegument proteins.
  • a virus-like particle of the invention comprises:
  • g. a combination of one or more capsid proteins; one or more tegument proteins and one or more surface proteins from different CMV strains;
  • h a combination of one or more capsid proteins, one or more tegument proteins, and one or more surface proteins from one or more CMV strains with additional T-cell epitopes;
  • capsid proteins i. a combination of one or more capsid proteins, one or more tegument proteins, and one or more surface proteins from one or more CMV strains excluding proteins responsible for immune evasion;
  • k a combination of one or more capsid proteins, one or more tegument proteins, and one or more surface proteins from one or more CMV strains further comprising proteins and/or motives enhancing the immune response, selected from the group of CD40L, CD19L, cytokines and unmethylated CpG motives;
  • I a combination of one or more capsid proteins, one or more tegument proteins, and one or more surface proteins from one or more CMV strains with proteins facilitating purification and analysis, selected e.g. from the group consisting of His- tag, Strep-tag, myc-tag, Flag-tag, and Snap-tag, and fluorescence proteins, selected e.g. from the group consisting of GFP, YFP and Red-cherry;
  • m. a combination of one or more capsid proteins, one or more tegument proteins, and one or more surface proteins from one or more CMV strains comprising further proteinaceous or non-protein structures to couple chemicals and/or inert nanobeads.
  • the strategy presented in the current invention to obtain large and complex CMV VLPs includes co-expression of multiple genes from single vectors, including genes for CMV capsid, tegument and/or surface proteins, as well as co-infection of several of these co- expression vectors.
  • rePAX recombinant assembly technology
  • the production of these novel multicomponent virus-like particles, protein complexes and soluble proteins is based on the recombinant assembly technology (known as rePAX) which includes the following steps: the assembly of genes, the generation of expression constructs for various systems, the protein expression, purification and characterization of the product.
  • the preferred expression system for the platform is the baculovirus system (BEVS) or the mammalian cell system. In another embodiment of the present invention a combination of both systems, known as BacMam, is used.
  • the herein described protein compositions such as VLPs, protein complexes and single proteins are generated in a shorter time and in unlimited amounts, due to the use of specific genetic and process engineering tools.
  • the use of these technologies does not require any physical transfer of original, potentially dangerous infectious or carcinogenic material during the development, manufacturing or administration of virus-like particles, protein complexes, and soluble proteins.
  • the vaccine of the invention is safe.
  • nucleotide sequences from HCMV available at the NCBI database.
  • the DNA encoding the proteins forming the VLPs of the invention, and vectors, either viral or plasmid based, comprising such DNA, are also part of the invention.
  • the vector backbone pRBT136 containing elements for propagation in E.coli and yeast (e.g. Saccharomyces cerevisae.) and for protein expression in insect cells are preferably used.
  • the vector backbone pRBT393 containing elements for gene assembly, homologous recombination and for expression in mammalian cells, preferably HEK293, CHO, human foreskin fibroblasts (HFF) and epithelial cells, are preferably used.
  • the parallel assembly of the different genes is conducted by homologous recombination of PCR products with homologous flanking regions and containing expression cassettes (promoter - gene of interest - terminator) or single genes fused together with protease cleavage sites such as the foot-and-mouse-disease virus 2A cleavage site into one open reading frame (ORF).
  • expression cassettes promoter - gene of interest - terminator
  • ORF open reading frame
  • the promoters are preferably selected from the group consisting of polh, p10 and p X
  • the terminators are preferably selected from the group consisting of SV40, HSVtk and BGH (bovine growth hormone).
  • the vector backbone pRBT136 used preferably for this invention contains an origin of replication for E.coli, e.g. pBR322ori, and yeast, e.g.2 micron ori, the polh and p10 promoters for expression in insect cells, the terminators SV40 and HSVtk, several resistance markers (ampicillin, gentamycin), a yeast selection marker (URA3), transposon sites (Tn) and a multiple cloning site (MCS).
  • E.coli e.g. pBR322ori
  • yeast e.g.2 micron ori
  • the polh and p10 promoters for expression in insect cells the terminators SV40 and HSVtk
  • several resistance markers ampicillin, gentamycin
  • UUA3 yeast selection marker
  • Tn transposon sites
  • MCS multiple cloning site
  • An expression cassette containing promoter - gene of interest - terminator is PCR amplified at the 5' site with a 35-40nt overhang at the 5' site, and at the 3' site with a further and different 35-40nt overhang.
  • the PCR product contains, at the 5' site, the complementary sequence of the 35-40nt overhang to the 3' site of the previous PCR product.
  • the remaining overhangs at the 5' site of the first PCR product and the 3' site of the second PCR product are homologous to the 3' and the 5' end of a linearized vector (pRBT136), respectively.
  • the homologous recombinations in a sequence are then conducted in yeast, preferably in Saccharomyces cerevisiae.
  • the number of the expression cassettes/PCR products to be assembled in parallel with the strategy described before is increased according to the needed number of genes to be assembled. By this means multiple genes / expression cassettes are assembled in parallel.
  • the assembled genes are flanked by the transposon sites. These are used for transposition of the genes into the baculovirus genome.
  • the resulting baculovirus co-expression vector ensures that the genes are co-expressed from the same single cell. Yield and product composition vary dependent on the number of proteins and production parameters.
  • the production parameters such as cell line, cell count at infection (CCI), amount of recombinant virus inoculum (multiplicity of infection, MOI) and time of harvest (TOH) are determined in respect to yield and early harvest using a matrix system and a small scale production system (2-20 ml; Ries, C, John C, Eibl R. (201 1 ), A new scale down approach for the rapid development of Sf21/BEVS based processes - a case study.
  • Eibl R., Eibl D. Editors: Single-use technology in Biopharmaceutical Manufacture, 207-213, John Wiley & Sons, Hoboken, New Jersey).
  • the defined parameters are then used to produce the respective product at larger scale.
  • Particles of the invention are manufactured using modern disposable tissue culture techniques which allow for high production capacity.
  • the preferred production cell lines of this invention are insect cell lines such as Sf9, Sf21 , Hi-5, Vankyrin (VE-1 , VE-2, VE-3), Express Sf+, and S2 Schneider cells.
  • insect cell lines such as Sf9, Sf21 , Hi-5, Vankyrin (VE-1 , VE-2, VE-3), Express Sf+, and S2 Schneider cells.
  • human cells e.g. HEK293, CHO, HeLa, Huh7, HepG2, BHK, MT-2, human foreskin fibroblasts (HFF), bone-marrow fibroblasts, primary neural cells, or epithelial cells are used.
  • yeast S. cerevisiae S. pombe, C. albicans, or P. pastoris cells are used.
  • Cultivation and propagation of host cells according to the invention is done in any vessel, bioreactor or disposable unit providing the appropriate conditions for the particular host cell.
  • CMV particles are purified using ultracentrifugation based classical glycerol-tartrate and sucrose gradients or modern chromatography based techniques, preferably affinity (IMAC) and ion exchange chromatography using large porous matrices such as membrane adsorbers and monoliths.
  • IMAC affinity
  • ion exchange chromatography using large porous matrices such as membrane adsorbers and monoliths.
  • the virus-like particles of the invention are used as therapeutics and/or as prophylactic vaccines. Furthermore they are used as antigens in diagnostic tools and as antigens for antibody generation. For both types of vaccines, either prophylactic or therapeutic, the antigen specific CD8+, cytotoxic T cell response as well as the antigen specific CD4+, T helper cell response is most important. Therefore the protein compositions of the VLPs are adapted to induce T- cell by expressing, e.g., pUL83, a known T-cell stimulator. In order to further address the different requirements of preventive vaccines, the present invention contains combinations of VLPs, soluble protein complex and single proteins.
  • compositions of the VLPs are adapted to contain immunogenic glycoproteins (gpUL75, gpUL55) and proteins necessary for formation of distinct complexes (gpUL1 15, gpUL128, gpUL130, gpUL131A).
  • immunogenic glycoproteins gpUL75, gpUL55
  • proteins necessary for formation of distinct complexes gpUL1 15, gpUL128, gpUL130, gpUL131A.
  • These combinations comprise the combination of soluble pentameric complex [gpUL75 (gH), gpUL1 15 (gL), gpUL128, gpUL130, gpUL131A] and virus-like particles, combination of one or more single soluble protein (such as pUL83, gpUL55) and the virus-like particles, and combination of the virus-like particles, the pentameric complex and a single soluble protein.
  • glycoproteins gpUL75 (gH), gpUL1 15 (gL) and gpUL55 (gB) are essential targets and are therefore expressed on the surface of the CMV-VLPs, preferably of the HCMV-VLPs.
  • a further aspect of this invention is the use of CMV-VLP and soluble pentameric complex (gpUL75 (gH), gpUL1 15 (gL), gpUL128, gpUL130, gpUL131A) in a prime-boost regimen, preferably priming with the VLPs and boosting with the pentameric complex. Priming with the pentameric complex and VLP boost is a suitable vaccination scheme.
  • CMV-VLP and soluble pentameric complex gpUL75 (gH), gpUL1 15 (gL), gpUL128, gpUL130, gpUL131A
  • VLP and soluble protein and/or VLP and DNA in such a prime-boost scenario is another aspect of this invention.
  • the invention relates to a vaccine comprising a recombinant virus-like particle as described, in particular to such a vaccine further comprising the pentameric complex consisting of gpUL75, gpUL1 15, gpUL128, gpUL130 and gpUL131A, and/or soluble CMV protein selected from the group consisting of gpUL75, gpUL1 15, gpUL55, gpUL74, gpUL100, and gpUL73; or gpUL128, gpUL130, and gpUL131A; or pUL83, IE-1 , UL99, UL91 , and pp150.
  • Such a vaccine may further comprise an adjuvant selected from the group consisting of aluminium hydroxide, alum, AS01 , AS02, AS03, AS04, MF59, MPL, QS21 , ISCOMs, IC31 , unmethylated CpG, ADVAX, RNA containing formulations and Freund's reagent, but also DNA of the invention as defined above.
  • an adjuvant selected from the group consisting of aluminium hydroxide, alum, AS01 , AS02, AS03, AS04, MF59, MPL, QS21 , ISCOMs, IC31 , unmethylated CpG, ADVAX, RNA containing formulations and Freund's reagent, but also DNA of the invention as defined above.
  • an adjuvant selected from the group consisting of aluminium hydroxide, alum, AS01 , AS02, AS03, AS04, MF59, MPL, QS21 , ISCOMs, IC31 , unmethylated CpG, AD
  • a method for vaccinating a human uses a pharmaceutical composition of the present invention comprising VLPs in the range of 10 ⁇ g to 10 mg/dose.
  • An average human of 70 kg is assumed to receive at least a single vaccination.
  • a dosage regimen comprising 3 doses applied at 0, 8 and 24 weeks, optionally followed by a second vaccination round 12-24 months after the last immunization is chosen.
  • Preferred routes of administration are subcutaneous (sc) and intramuscular (im) administration, but intradermal and intranasal are also suitable administrations.
  • compositions of the VLPs of this invention are likewise used in a method of therapeutic treatment.
  • a method for vaccinating a human for treatment purposes uses a pharmaceutical composition of the present invention comprising VLPs in the range of 10 ⁇ g to 10 mg/dose.
  • An average human of 70 kg is assumed to receive at least a single vaccination.
  • a dosage regimen comprising 3 doses applied at 0, 8 and 24 weeks, optionally followed by a second vaccination round 12-24 months after the last immunization is chosen.
  • the pharmaceutical composition of the present invention comprises VLPs in the range of 5 ⁇ g to 10mg/dose for women and half this dose for children.
  • the pharmaceutical compositions comprise VLP, pentameric complex, and/or soluble protein as separate or combined pharmaceutical compositions for a prime-boost regimen.
  • Each component is used in the range of 10 ⁇ g to 10 mg/dose, respectively.
  • the pharmaceutical composition of the combination of VLP and pentameric complex and/or soluble protein is in the range of 10 ⁇ g to 10 mg/dose of each component. Different ratios of the components are likewise possible.
  • An average human of 70 kg is assumed to receive at least one single vaccination.
  • a dosage regimen comprising 3 doses applied at 0, 8 and 24 weeks, optionally followed by a second vaccination round 12-24 months after the last immunization is chosen.
  • Preferred routes of administration are subcutaneous (sc) and intramuscular (im) administration, but intradermal and intranasal are also suitable administrations.
  • the method of treatment of the invention is particularly important for solid organ, bone marrow and/or stem cell transplant recipients. For these patients a fast and effective CD8+ and CD4+ T-cell response is crucial.
  • the pharmaceutical composition of the present invention is in the range of 10 ⁇ g to 10 mg/dose.
  • An average human of 70 kg is assumed to receive at least once a vaccination.
  • a dosage regimen comprising 3 doses applied at 0, 2 and 4 weeks before transplantation, optionally followed by a second vaccination round 1 , 4, 8 weeks after the transplantation is chosen.
  • Vaccines according to the invention comprise the recombinant virus-like particle, the pentameric complex and/or the soluble protein in aqueous solution, and optionally further viscosity-regulating compounds, stabilizing compounds and/or an adjuvant increasing the immunogenicity, as it is known in the state of the art.
  • Example 1 Construction of recombinant expression vectors RBT136-7 and RBT136-9
  • the vector backbone pRBT136 containing elements for propagation in E.coli and yeast (e.g. Saccharomyces cerevisae) and for protein expression in insect cells, is used.
  • yeast e.g. Saccharomyces cerevisae
  • protein expression in insect cells is used.
  • different genes were chosen for providing the capsid, the tegument and surface proteins.
  • the genes are generated by gene synthesis with a specific optimization for Spodoptera frugiperda and chosen out of the following groups of the cytomegalovirus genes: first from the capsid compartment, e.g. pUL86, pUL85, pUL46, pUL48, pUL50, and pUL80.5 (UL80A); second from the tegument part, e.g. pUL83, pUL25, ppUL32, and pUL99, and third from the surface part, e.g.
  • gpUL55 (gB), gpUL75 (gH), gpUL1 15 (gl_), gpUL74 (gO), gpUU OO (gM), gpUL73 (gN), gpUL128, gpUL130, and gpUL131A.
  • pUL86, pUL85, pUL46, and pUL80a optionally in combination with the functional genes pUL48a and pUL50, were used.
  • the tegument chosen is pUL83 whereas the surface was prepared from gpUL55 (gB), gpUL75 (gH), gpUL1 15 (gl_), gpUL74 (gO), gpUL128, gpUL130, and gpUL131A.
  • the vector backbone pRBT136 contains an origin of replication for E.coli and yeast, the polh and p10 promoters for expression in insect cells, the terminator SV40 and HSVtk, several resistance markers (ampicillin, gentamycin), transposon sites (Tn) and a multiple cloning site (MCS).
  • the expression cassette containing promoter - UL86 - terminator was PCR amplified at the 5' site with the primer RBT155 (TG ATTTG ATAATAATTCTTATTTAAC , SEQ ID NO:63) and at the 3' site with a 35-40nt overhang (G GCTAG CTTTGTTTAACTTTAAG AAG GAG ATACATCTAG A, SEQ ID NO:64).
  • homologous recombination with the expression cassette containing promoter - UL85 - terminator, this part was PCR amplified at the N-terminal end with the complementary sequence of the 35-40nt overhang at the C-terminal end of the previous PCR product.
  • the homologous recombination took place in yeast (Saccharomyces cerevisiae): An overnight culture was centrifuged at 500xg at 25°C for 5 min, the supernatant was aspirated and the pellet washed with water and with Tris-EDTA-LiOAc buffer.
  • the vector, linearized with EcoRI and Hind 111 (1 ⁇ ), salmon sperm DNA (8 ⁇ ) and the PCR products (1 -4 ⁇ ) representing the expression cassettes were added to 50 ⁇ washed yeast, incubated for 30 min at 30°C, followed by a heat shock at 42°C, and plated on a respective amino acid drop out agar plate without uracil, and incubated for 3 days.
  • the resulting vector was isolated from the yeast cell by freeze (liquid nitrogen, 2 min) - thaw (95°C, 1 min) - lysis. After addition of the same volume chloroform the vector was precipitated with the double volume of ethanol.
  • the precipitated vector (5 ⁇ ) was added to 50 ⁇ competent XL-1 blue (Stratagene) followed by a transformation according to the manufacturer's protocol: Incubation on ice for 30 min, followed by a heat shock at 42°C for 45 sec, a 2 min cold shock at 4°C, addition of 200 ⁇ 2YT medium, and a 1 h incubation at 220 rpm at 37°C.
  • the culture was then plated on 2YT agar plates containing 100 ⁇ g ampicillin and 100 ⁇ g gentamycin. The grown clones were verified by PCR screening, analytical digests and sequencing to prove the presence of all assembled genes.
  • the genes UL83 and UL80.5 were assembled into the vectors pRBT4 and combined by cre-lox recombination with pRBT136 containing pUL86-pUL85-pUL50-pUL48-pUL46- gpUL74 (gO). 1 ⁇ cre-recombinase was added to 500 ng of pRBT5 and pRBT136 in a 10 ⁇ total volume and incubated for 30 min at 37°C. After a 10 min heat inactivation at 70°C a transformation in XL-1 blue competent cells was performed as described above. The clones were verified by PCR screening, analytical digest and sequencing. The sequenced plasmids were used for the generation of the corresponding baculovirus genome (bacmid).
  • the genes of interest were transferred by site-specific homologous recombination via Tn7 directed transposition into competent DH10MB cells.
  • 10 ng sequenced plasmid was added to 100 ⁇ competent DH10MB cells and incubated for 30 min at 4°C. After a heat shock at 42°C for 45 sec, a 2 min cold shock at 4°C, addition of 400 ⁇ 2YT medium and a 4 h incubation at 37°C at 220 rpm, a 1 :10 dilution was plated on appropriate 2YT agar plates containing the antibiotics according to the resistance markers and IPTG/X-gal for blue-white screening.
  • the infective titer of the working seed virus was determined by a classical plaque assay. Virus dilutions (10 5 to 10 8 in a volume of 1 ml) were added to 1 x10 6 cells/6-well Sf9 cell culture, and incubated for 1 h at 27°C. After aspiration of the virus, the cells were overlayed with 4% agarose in Sf900-1.3 medium, followed by an incubation at 27°C in a humidified chamber. The infective titer of the different constructs were in the range of 1 -5x10 7 pfu/cell.
  • the production parameters such as cell line, cell count at infection (CCI), amount of recombinant virus inoculum (multiplicity of infection, MOI) and time of harvest (TOH) were determined in 50 ml bioreactors (Cultiflask, Sartorius Stedim) by infection of a 20 ml insect cell culture.
  • CCI cell count at infection
  • MOI multiplicity of infection
  • TOH time of harvest
  • c consensus sequence
  • H His-tag
  • SH Streptavidin-His-tag
  • V VR1814
  • pel precission protease
  • pell precission and TEV protease
  • DT dimerization tool
  • Example 2 Expression vectors pRBT136CMV-1 ; pRBT136CMV-8 and pRBT136CMV-20
  • each gene of interest namely UL86, UL55 (gB), and gag
  • BamHI 5'
  • Hindlll 3'
  • the ligation was conducted with 2 ⁇ g cut pRBT136, 10 ⁇ g fragment and 1 ⁇ T4-ligase (NEB) in a total volume of 20 ⁇ , followed by a transformation in competent XL-1 blue cells as described in Example 1 .
  • Example 3 Expression construct for capsid variant pRBTCMV-2
  • the virus-like particles (VLP) comprising UL86-UL85-UL50-UL48-UL46-UL83- UL80.5-UL74- gH-gL-UL128-UL130-UL131A are produced in disposable 1 L shake flasks (culture volume 300 mL) in fall army worm Spodoptera frugiperda cells (Sf9) by co- infection of 2 baculoviruses (SEQ ID NO:5 and SEQ ID NO:12) containing each multiple genes.
  • the production parameters were as follows: Initial cell count at infection (CCI) of 2x10 6 cells/ml, a multiplicity of infection (MOI) of 0.4 pfu/ml, for each virus 0.2 pfu/ml, incubation at 27°C at 100 rpm. Harvest took place at day 3 post infection (p.i.) at a viability around 80%. The production was controlled by daily sampling, determining cell count and viability.
  • CCI initial cell count at infection
  • MOI multiplicity of infection
  • the VLP containing supernatant was concentrated 3x using tangential-flow- filtration with a Hydrosart Sartocon Slice200 cassette with a molecular weight cut off (MWCO) of 100 kDa and a filter area of 0.02 m 2 (Sartorius Stedim) at a flow rate of 80 ml/min and a transmembrane pressure between 1 -1 .5 bar.
  • MWCO molecular weight cut off
  • the retentate was subjected to purification by a 2-step glycerol-tartrate gradient ultracentrifugation.
  • retentate were overlayed onto a 6 mL 30% sucrose cushion (w/v in 50 mM Tris, 100 mM NaCI, pH 7.4) and pelleted for 1.5 h at 16000 rpm and 16°C using a SW28 rotor.
  • the pellet was resuspended in 0.5 ml TN-buffer (50 mM Tris, 100 mM NaCI, pH 7.4).
  • a glycerol-tartrate gradient (30% glycerol mixed with 15-35% tartrate, 12 ml volume) was prepared, overlayed with the 0.5 ml pre-purified material and centrifuged for 16 h at 24000 rpm and 16°C using a SW41 rotor. The gradient was fractionated from top in 0.9 ml fractions and the purified VLPs were analysed by biochemical methods. 150 ⁇ of different gradient fractions were loaded onto a 4-12% Bis-Tris NuPAGE gel (Invitrogen) according to the manufacturer's protocol, run for 15 min at 150 V and for 45 min at 180 V using MOPS running buffer. The gels were stained over night with SimplyBlue SafeStain reagent (Invitrogen) and destained with water.
  • the different gradient fractions were 1 :10 diluted in 100 ⁇ /well coating buffer (0.1 M Na 2 HP0 4 , pH 9) in a 96-well pre-absorbed ELISA plate and incubated over night at 4°C. Afterwards the plate was washed 3x with 195 ⁇ /well wash buffer (1 xPBS, 0.05% Tween 20) followed by a 1 h blocking at room temperature with 195 ⁇ /well 3%BSA in 1x PBS solution.
  • the specific antibody the anti-surface antibody (anti-gH,) as well as the anti-tegument antibody (anti-UL83) in a concentration of 1 ⁇ glm ⁇ in 3%BSA, 1 x PBS, 0.05% Tween 20, pH 7 was added
  • Table 2 is an overview of the CMV-VLP variants which were expressed, purified and characterized as described in Example 5.
  • the genetic constructs described in Table 1 were quality controlled by sequencing before they were used for further manipulation such as generation of the corresponding baculoviruses.
  • the integrity of the generated baculoviruses is quality controlled by PCR based transgene control for presence of each single gene.
  • the most important properties for manufacturing and regulatory approval such as yield and co-localization of the proteins, especially of the immunogenic surface proteins of these CMV VLPs are stated in Table 2.
  • Example 6 Expression, purification and characterization of the pentameric CMV complex.
  • the pentameric CMV complex comprising gpUL75 (gH-His)- gpUL1 15 (gL)-gpUL128-gpUL130-gpUL131A is produced in disposable 2 L shake flasks (culture volume 700 ml) in fall army worm Spodoptera frugiperda cells (Sf9) by co- expression from a single baculovirus (SEQ ID NO:59).
  • the production parameters were as follows: Initial cell count at infection (CCI) of 2x10 6 cells/ml, a multiplicity of infection (MOI) of 0.25 pfu/ml, incubation at 27°C at 100 rpm. Harvest took place at day 3 post infection (p.i.) at a viability around 80%. The production was controlled by daily sampling, determining cell count and viability.
  • the complex containing supernatant was loaded on 2x 5 ml HisTrap colums (GE Healthcare). The complex was purified using a linear gradient from zero to 500 mM imidazole over 50 column volumes (CV, equivalent to 500 ml. The different chromatographic fractions were analysed by biochemical methods.
  • Total protein is determined via a Bradford assay adapted to a 96-well plate format (BCA, Pierce). 20 ⁇ of unknown or standard sample was diluted in 180 ⁇ of buffer. Serial 2-fold dilutions are made to the standard in triplicate (pure bovine serum albumin) or unknown samples. 100 ⁇ of 2x stock Bradford reagent (Pierce) was added per well. The plate is then mixed and absorbance was measured at 595 nm in a microtiter plate reader.
  • the pentameric CMV complex contained in the different fractions from the chromatographic based purification is further analysed by investigating the binding of a specific antibody against the His-tag added to gpUL75 (gH, mouse-anti-His, AbD Serotec) and the gpUL75 itself (gH, mouse-anti-gH, Santa Cruz). Therefore the different samples were 1 :10 diluted in 100 ⁇ l/well coating buffer (0.1 M Na 2 HP0 4 , pH 9) in a 96-well pre-absorbed ELISA plate and incubated over night at 4°C.
  • the plate was washed 3x with 195 ⁇ /well wash buffer (1 xPBS, 0.05% Tween 20) followed by a 1 h blocking at room temperature with 195 ⁇ /well 3%BSA in 1x PBS solution.
  • the specific antibody, the anti-His antibody (anti-His) as well as the anti-gpUL75 (anti-gH) in a concentration of 1 g/ml in 3%BSA, 1x PBS, 0.05% Tween 20, pH 7 was added (100 ⁇ /well) and incubated for 1 h at room temperature followed by further 3 wash steps.
  • a 1 h incubation with the appropriate secondary antibody (anti-mouse-lgG-HRP, 1 :1000 dilution in 3%BSA, 1x PBS, 0.05% Tween20, pH 7) was conducted.
  • the binding of the specific antibody to the pentameric complex was detected using 100 ⁇ /well TMP substrate reagent (BD Biosciences, San Diego, USA; according to manufacturer ' s protocol), wherefore the reaction is stopped after 3-15 min with 100 ⁇ 1 M HCI, followed by OD measurement at 450 nm in a microplate reader.
  • VLP sample was incubated 10 min on a copper covered EM grid, stained with 5 ⁇ phosphor tungsten acid for 5-10 min and de-stained 2 times with water for 1 min.
  • the liquid on the grid was removed with a Whatman paper and the grid was transferred to the JEOL3000 microscope, investigated and pictures of the particles taken.
  • Example 8 Binding assays by the use of sandwich ELISA
  • a sandwich ELISA as binding assay was performed to determine the presence and accessibility of the surface proteins of different reVLPs (recombinant VLPs). Therefore a specific surface and/or tegument antibody in a concentration of 1 ⁇ g ml in 3%BSA, 1 x PBS, 0.05% Tween 20, pH 7 was added (100 ⁇ /well) in coating buffer (0.1 M Na 2 HP0 4 , pH 9) in a 96-well pre-absorbed ELISA plate and incubated over night at 4°C.
  • the plate was washed 3x with 195 ⁇ /well wash buffer (1 xPBS, 0.05% Tween 20) followed by a 1 h blocking at room temperature with 195 ⁇ /well 3% BSA in 1x PBS solution.
  • the different VLPs in a 1 :10 dilution in 3% BSA, 1 x PBS, 0.05% Tween 20, pH 7 were added (100 ⁇ /well) and incubated for 1 h at room temperature.
  • the specific antibody, the anti-surface antibody (anti-gH, anti-gB) as well as the anti- tegument antibody (anti-UL83) in a concentration of 1 ⁇ / ⁇ in 3% BSA, 1 x PBS, 0.05% Tween 20, pH 7 were added (100 ⁇ /well) and incubated for 1 h at room temperature followed by further 3 wash steps.
  • the appropriate secondary antibody anti-mouse-lgG-HRP, 1 :1000 dilution in 3% BSA, 1 x PBS, 0.05% Tween 20, pH 7 was conducted.
  • the binding of the specific antibody to the VLP was detected using 100 ⁇ /well TMP substrate reagent (BD Biosciences, San Diego, USA; according to manufacturer ' s protocol). The reaction is stopped after 3-15 min with 100 ⁇ 1 M HCI, followed by OD measurement at 450 nm in a microplate reader.
  • mice were used in a prime-boost-boost regimen. Each group contained 8 mice and each mouse received 20 ⁇ g protein per injection. For the combination (CMV-reVLP + pentameric complex) a total amount of 40 ⁇ g protein was injected. Pre-immune sera were taken after 14 days quarantine of the mice (day 0). The first injection took place 10 days later followed by a booster injection at day 42. The first bleeding was done at day 49. The 2 nd booster injection was performed at day 61 followed by a further bleeding at day 70. The final bleeding took place at day 85 followed by the investigation of humoral and cellular immune response.
  • CMV-reVLP + pentameric complex Pre-immune sera were taken after 14 days quarantine of the mice (day 0). The first injection took place 10 days later followed by a booster injection at day 42. The first bleeding was done at day 49. The 2 nd booster injection was performed at day 61 followed by a further bleeding at day 70. The final bleeding took place at day 85 followed by the investigation of humor
  • Example 10 Humoral immune response based on neutralization assay.
  • the humoral immune response of the vaccine candidate based on the combination of a CMV-reVLP (vRBT-20) and the pentameric complex (SEQ ID: 59) was investigated by a neutralization assay of the mice sera from example 9 in comparison to sera from CMV negative and CMV positive human blood donors.
  • a BAC (bacterial artificial chromosome)- reconstituted VR1814 strain carrying a GFP molecule for analytical reasons (fix-EGFP) was used for the infection of fibroblasts (MRC-5) and epithelial (ARPE-19) cells to visualize the neutralisation potential of the mouse sera from Example 9. 2x10 4 cells / well were seeded into a 96 well plate in RPMI medium containing 10% FCS (fetal calf serum).
  • a serum pool of the 8 mice was generated and added in 2-fold serial dilutions (1 :20 to 1 :2560) in 100 ⁇ l well RPMI/FCS medium.
  • the above mentioned fix-EGFP VR1814 virus was added in a tissue culture infectious dose (TCID) of 1000 virus molecules/well which was determined in a pre-assay.
  • TCID tissue culture infectious dose
  • the 96 well plates were incubated for 8 days at 37°C in a C0 2 controlled atmosphere.
  • the determination of green cell was performed in a plate reader with the following parameters: fluorescein-filter [excitation 485/20, emission 530/25), bottom reading mode, time: 0.1 sec, 25x measurements/well after 96 h incubation.
  • the neutralization potency was determined as the dilution of the sera able to show a 50% virus infection inhibition.
  • the following controls were performed: cell control (cells + PBS), virus control (only infected cells) and 5 CMV positive and 5 CMV negative sera from human blood donors.
  • Example 1 1 Cellular immune response based on EliSpot data (Multiplex assay).
  • mice were killed and the spleenocytes prepared for the analysis of 10 different cytokines.
  • the spleenocytes were prepared for the analysis of 10 different cytokines.
  • CMV-reVLP as well as mixes of synthetic peptides were used.
  • the following proteins were verified: HIVgag, pUL83, gpUL75, gpUL1 15, gpUL55, gpUL128, gpUL130 and gpUL131A.
  • cytokines IFN-gamma, IL-1 beta, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, gCMSF and TNFalpha
  • IFN-gamma IFN-gamma
  • IL-1 beta IL-1 beta
  • IL-2 IL-2
  • IL-4 IL-5
  • IL-6 IL-6
  • IL-10 IL-12
  • gCMSF TNFalpha
  • anti-CMV monoclonal antibodies such as anti-gH, anti-gB, anti-UL83, anti-UL86, anti-UL85, anti-UL130/UL131A, anti-gag, anti-His were diluted to a final concentration of ⁇ g/ml coating buffer (0.1 0.1 M Na 2 HP0 4 , pH 9) in a 96 well pre-absorbed ELISA plate (1 ⁇ /well) and incubated over night at 4°C.
  • the plate was washed 3x with 195 ⁇ /well wash buffer (1 x PBS, 0.05% Tween 20) followed by a 1 h blocking at room temperature with 195 ⁇ /well 3% BSA in 1 x PBS solution.
  • the different CMV VLPs and soluble complex in a 1 :10 dilution in 3% BSA, 1 x PBS, 0.05% Tween 20, pH 7 were added (100 ⁇ /well) and incubated for 1 h at room temperature.
  • the reaction is stopped after 3- 15 min with 100 ⁇ 1 M HCI, followed by OD measurement at 450 nm in a microplate reader.
  • This kind of sandwich assay was chosen to verify the integrity of the CMV VLPs and the soluble complex based on the binding on one side to monoclonal antibodies and on the other side to antibodies present in CMV positive sera. The binding to human antibodies in the sera showed the accessibility of proteins of the CMV VLPs and soluble complex.
  • Example 13 Characterization of different CMV-reVLPs and the soluble complex using blood serum from CMV positive donors
  • the different CMV-reVLP variants were produced as described in Example 5 and subjected to concentration by a sucrose cushion based ultracentrifugation. 25 ml cell culture supernatant was overlayed onto a 6 mL 30% sucrose cushion (w/v in 50 mM Tris, 100 mM NaCI, pH 7.4) and pelleted for 1.5 h at 20'000 rpm and 16°C using a SW28 rotor. The pellet was resuspended in 0.2-0.4 ml TN-buffer (50 mM Tris, 100 mM NaCI, pH 7.4). 50 ⁇ of the resuspended pellet was loaded onto a 4-12% Bis-Tris NuPAGE gel

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)

Abstract

La présente invention concerne des ensembles de gènes et de protéines sous forme de particules de type viral et des complexes de protéines pour utilisation en tant que vaccins prophylactiques ou thérapeutiques, et des outils de diagnostic et de R & D pour le cytomégalovirus humain (HCMV) et d'autres herpèsvirus. Les particules de type viral comprennent une ou plusieurs protéines de capside d'un herpèsvirus ou un rétrovirus, trois protéines de surface de CMV ou plus et éventuellement les protéines de tégument. Les ensembles sont préparés en utilisant une technologie combinant de l'ADN recombinant avec des techniques de culture de cellules et de purification disponibles.
PCT/EP2013/072717 2012-10-30 2013-10-30 Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain WO2014068001A1 (fr)

Priority Applications (15)

Application Number Priority Date Filing Date Title
EP13789738.5A EP2914284B1 (fr) 2012-10-30 2013-10-30 Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain
CA2885145A CA2885145A1 (fr) 2012-10-30 2013-10-30 Vaccins recombinants a base de particules contre l'infection par le cytomegalovirus humain
SG11201501623PA SG11201501623PA (en) 2012-10-30 2013-10-30 Recombinant particle based vaccines against human cytomegalovirus infection
RU2015121828A RU2015121828A (ru) 2012-10-30 2013-10-30 Вакцины на основе рекомбинантных частиц против инфекции цитомегаловируса человека
CN201380057084.6A CN104853772A (zh) 2012-10-30 2013-10-30 抗人巨细胞病毒感染的基于重组颗粒的疫苗
AU2013340820A AU2013340820A1 (en) 2012-10-30 2013-10-30 Recombinant particle based vaccines against human cytomegalovirus infection
JP2015540112A JP6294890B2 (ja) 2012-10-30 2013-10-30 ヒトサイトメガロウイルス感染に対する組換え粒子ベースのワクチン
ES13789738.5T ES2608637T3 (es) 2012-10-30 2013-10-30 Vacunas basadas en partículas recombinantes contra la infección por el citomegalovirus humano
BR112015008930A BR112015008930A2 (pt) 2012-10-30 2013-10-30 vacinas baseadas em partícula recombinante contra a infecção por citomegalovírus humano
KR1020157013906A KR20150076244A (ko) 2012-10-30 2013-10-30 인간 시토메갈로바이러스 감염에 대한 재조합 입자 기재 백신
MX2015005505A MX2015005505A (es) 2012-10-30 2013-10-30 Vacunas basadas en particular recombinantes contra la infeccion de citomegalovirus humano.
US14/436,943 US20150359879A1 (en) 2012-10-30 2013-10-30 Recombinant particle based vaccines against human cytomegalovirus infection
IL237793A IL237793A0 (en) 2012-10-30 2015-03-16 Vaccines against human cytomegalovirus infection based on recombinant particles
PH12015500931A PH12015500931A1 (en) 2012-10-30 2015-04-24 Recombinant particle based vaccines against human cytomegalovirus infection
HK16101176.6A HK1213185A1 (zh) 2012-10-30 2016-02-02 抗人巨細胞病毒感染的基於重組顆粒的疫苗

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12190652 2012-10-30
EP12190652.3 2012-10-30

Publications (1)

Publication Number Publication Date
WO2014068001A1 true WO2014068001A1 (fr) 2014-05-08

Family

ID=47073362

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/072717 WO2014068001A1 (fr) 2012-10-30 2013-10-30 Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain

Country Status (16)

Country Link
US (1) US20150359879A1 (fr)
EP (2) EP2914284B1 (fr)
JP (1) JP6294890B2 (fr)
KR (1) KR20150076244A (fr)
CN (1) CN104853772A (fr)
AU (1) AU2013340820A1 (fr)
BR (1) BR112015008930A2 (fr)
CA (1) CA2885145A1 (fr)
ES (1) ES2608637T3 (fr)
HK (1) HK1213185A1 (fr)
IL (1) IL237793A0 (fr)
MX (1) MX2015005505A (fr)
PH (1) PH12015500931A1 (fr)
RU (1) RU2015121828A (fr)
SG (1) SG11201501623PA (fr)
WO (1) WO2014068001A1 (fr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015161926A1 (fr) * 2014-04-23 2015-10-29 Institute For Research In Biomedicine Compositions de vaccin contre le cytomégalovirus humain et méthode de production associée
WO2015165480A1 (fr) * 2014-04-30 2015-11-05 Institute For Research In Biomedicine Compositions de vaccin de cytomégalovirus humain et leur procédé de production
WO2015170287A1 (fr) * 2014-05-08 2015-11-12 Redvax Gmbh Moyens et procédés de traitement de cmv
WO2017070613A1 (fr) 2015-10-22 2017-04-27 Modernatx, Inc. Vaccin contre le cytomégalovirus humain
WO2017157437A1 (fr) 2016-03-16 2017-09-21 Institut Curie Procédé de préparation de particules virales à dinucléotide cyclique et utilisation desdites particules pour le traitement du cancer
JP2018504117A (ja) * 2015-01-22 2018-02-15 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム サイトメガロウイルス抗原およびその使用
US10010607B2 (en) 2014-09-16 2018-07-03 Institut Curie Method for preparing viral particles with cyclic dinucleotide and use of said particles for inducing immune response
JP2019088323A (ja) * 2014-07-16 2019-06-13 オレゴン ヘルス アンド サイエンス ユニバーシティ 外来抗原を含むヒトサイトメガロウイルス
FR3077490A1 (fr) * 2018-02-02 2019-08-09 Universite De Franche-Comte Nouvelles compositions vaccinales pour lutter contre le cancer du sein, et procede de preparation
US10611800B2 (en) 2016-03-11 2020-04-07 Pfizer Inc. Human cytomegalovirus gB polypeptide
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11629172B2 (en) 2018-12-21 2023-04-18 Pfizer Inc. Human cytomegalovirus gB polypeptide
US11857622B2 (en) 2020-06-21 2024-01-02 Pfizer Inc. Human cytomegalovirus GB polypeptide

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3060019A1 (fr) 2017-04-19 2018-10-25 Glaxosmithkline Biologicals Sa Proteines de cytomegalovirus modifiees et complexes stabilises
BR112020004747A2 (pt) * 2017-09-13 2020-09-24 Sanofi Pasteur composição imunogênica de citomegalovírus humano
EP3762025A4 (fr) * 2018-03-09 2021-12-01 The Regents of the University of California Vecteurs de cmv et utilisations associées
WO2019216929A1 (fr) * 2018-05-11 2019-11-14 City Of Hope Vecteurs mva pour exprimer de multiples antigènes du cytomégalovirus (cmv), et utilisation de ces derniers
KR102018341B1 (ko) * 2018-06-21 2019-09-04 성균관대학교산학협력단 RNF170 및 pUL50을 유효성분으로 포함하는 암 예방 또는 치료용 약학적 조성물
JP2022505094A (ja) 2018-10-17 2022-01-14 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 改変されたサイトメガロウイルスタンパク質及び安定化された複合体
WO2021108238A1 (fr) * 2019-11-26 2021-06-03 Merck Sharp & Dohme Corp. Procédé de détection du cytomégalovirus (cmv) et de mesure et de quantification d'un complexe pentamérique à l'aide d'un dosage elisa sandwich indirect
WO2023223255A1 (fr) 2022-05-20 2023-11-23 Glaxosmithkline Biologicals Sa Protéines de la glycoprotéine e du virus varicelle-zona modifiée

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138590A1 (fr) * 2007-05-11 2008-11-20 Vakzine Projekt Management Gmbh Composition contenant des particules de cytomégalovirus humain (cmvh)
WO2010125201A1 (fr) * 2009-05-01 2010-11-04 Redbiotec Ag Particules recombinantes de types virus codées par un vecteur de gènes multiples
WO2010128338A2 (fr) * 2009-05-08 2010-11-11 Henderson Morley Plc Vaccins

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5990085A (en) 1998-05-04 1999-11-23 Michigan State University Inhibin-HBc fusion protein
AU2005253723B2 (en) 2004-06-16 2011-01-20 Glaxosmithkline Biologicals S.A. Vaccine against HPV16 and HPV18 and at least another HPV type selected from HPV 31, 45 or 52
CA2728739C (fr) * 2008-06-20 2017-07-11 Duke University Utlisation d'antigenes du cytomegalovirus humain pour ameliorer les reponses immunitaires aux cellules cancereuses
CA2840989A1 (fr) * 2011-07-06 2013-01-10 Novartis Ag Compositions de combinaisons immunogenes et utilisations de celles-ci
CA2889659C (fr) * 2011-11-11 2023-03-14 Variation Biotechnologies Inc. Compositions et methodes pour le traitement d'infections par cytomegalovirus

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008138590A1 (fr) * 2007-05-11 2008-11-20 Vakzine Projekt Management Gmbh Composition contenant des particules de cytomégalovirus humain (cmvh)
WO2010125201A1 (fr) * 2009-05-01 2010-11-04 Redbiotec Ag Particules recombinantes de types virus codées par un vecteur de gènes multiples
WO2010128338A2 (fr) * 2009-05-08 2010-11-11 Henderson Morley Plc Vaccins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TANG XIAN-CHUN ET AL: "Baculovirus-Produced Influenza Virus-like Particles in Mammalian Cells Protect Mice from Lethal Influenza Challenge", VIRAL IMMUNOLOGY, vol. 24, no. 4, August 2011 (2011-08-01), pages 311 - 319, XP002711212 *

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2015161926A1 (fr) * 2014-04-23 2015-10-29 Institute For Research In Biomedicine Compositions de vaccin contre le cytomégalovirus humain et méthode de production associée
WO2015165480A1 (fr) * 2014-04-30 2015-11-05 Institute For Research In Biomedicine Compositions de vaccin de cytomégalovirus humain et leur procédé de production
WO2015170287A1 (fr) * 2014-05-08 2015-11-12 Redvax Gmbh Moyens et procédés de traitement de cmv
CN106460010A (zh) * 2014-05-08 2017-02-22 辉瑞公司 治疗cmv之手段及方法
JP2019088323A (ja) * 2014-07-16 2019-06-13 オレゴン ヘルス アンド サイエンス ユニバーシティ 外来抗原を含むヒトサイトメガロウイルス
US10010607B2 (en) 2014-09-16 2018-07-03 Institut Curie Method for preparing viral particles with cyclic dinucleotide and use of said particles for inducing immune response
JP2018504117A (ja) * 2015-01-22 2018-02-15 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム サイトメガロウイルス抗原およびその使用
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
WO2017070613A1 (fr) 2015-10-22 2017-04-27 Modernatx, Inc. Vaccin contre le cytomégalovirus humain
US10611800B2 (en) 2016-03-11 2020-04-07 Pfizer Inc. Human cytomegalovirus gB polypeptide
WO2017157437A1 (fr) 2016-03-16 2017-09-21 Institut Curie Procédé de préparation de particules virales à dinucléotide cyclique et utilisation desdites particules pour le traitement du cancer
EP3770265A1 (fr) 2016-03-16 2021-01-27 Institut Curie Procédé de préparation de particules virales à dinucléotide cyclique et utilisation desdites particules pour le traitement du cancer
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
FR3077490A1 (fr) * 2018-02-02 2019-08-09 Universite De Franche-Comte Nouvelles compositions vaccinales pour lutter contre le cancer du sein, et procede de preparation
US11629172B2 (en) 2018-12-21 2023-04-18 Pfizer Inc. Human cytomegalovirus gB polypeptide
US11857622B2 (en) 2020-06-21 2024-01-02 Pfizer Inc. Human cytomegalovirus GB polypeptide
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine

Also Published As

Publication number Publication date
SG11201501623PA (en) 2015-05-28
JP2015536937A (ja) 2015-12-24
CA2885145A1 (fr) 2014-05-08
BR112015008930A2 (pt) 2017-11-21
EP3269388A1 (fr) 2018-01-17
AU2013340820A1 (en) 2015-03-19
US20150359879A1 (en) 2015-12-17
PH12015500931A1 (en) 2015-06-29
CN104853772A (zh) 2015-08-19
JP6294890B2 (ja) 2018-03-14
EP2914284A1 (fr) 2015-09-09
MX2015005505A (es) 2016-01-08
IL237793A0 (en) 2015-05-31
KR20150076244A (ko) 2015-07-06
EP2914284B1 (fr) 2016-09-21
ES2608637T3 (es) 2017-04-12
RU2015121828A (ru) 2016-12-20
HK1213185A1 (zh) 2016-06-30

Similar Documents

Publication Publication Date Title
EP2914284B1 (fr) Vaccins recombinants à base de particules contre l'infection par le cytomégalovirus humain
Anderholm et al. Cytomegalovirus vaccines: current status and future prospects
US7163685B2 (en) Human cytomegalovirus antigens expressed in MVA and methods of use
JP3624911B2 (ja) 組換えアビポックスウイルス
US6713070B1 (en) Viral particles which are released after the infection with the human cytomegalovirus, and the use of said particles as a vaccine
EP3139953A1 (fr) Moyens et procédés de traitement de cmv
DK2556150T3 (en) A viral particle released after infection of mammalian cells by human cytomegalovirus (HCMV) containing a fusion protein and use thereof
ES2716010T3 (es) Métodos y composiciones para la proteína IL-10 de citomegalovirus
Gómez et al. Enhanced CD8+ T cell immune response against a V3 loop multi-epitope polypeptide (TAB13) of HIV-1 Env after priming with purified fusion protein and booster with modified vaccinia virus Ankara (MVA-TAB) recombinant: a comparison of humoral and cellular immune responses with the vaccinia virus Western Reserve (WR) vector
US11844833B2 (en) Expression system for expressing herpesvirus glycoprotein complexes
JP2024500167A (ja) 増加した免疫原性を有する改変パラポックスウイルス
US20220143174A1 (en) Multivalent kaposi sarcoma-associated herpesvirus-like particles and uses thereof
KR100263278B1 (ko) Alvac 카나리아폭스 바이러스 재조합체 및 그것의 사용
Morgan et al. The Development of Epstein–Barr Virus Vaccines
Wussow et al. Human Cytomegalovirus Vaccine Based on the Envelope gH/gL Pentamer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13789738

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2885145

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 237793

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2013340820

Country of ref document: AU

Date of ref document: 20131030

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14436943

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 12015500931

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 15098495

Country of ref document: CO

Ref document number: MX/A/2015/005505

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2015540112

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 000581-2015

Country of ref document: PE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015008930

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2013789738

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2013789738

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157013906

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: IDP00201503221

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 2015121828

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015008930

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150420